(FabZ) of Helicobacter pylori - American Chemical Society

Mar 23, 2009 - School of Pharmacy, East China University of Science and Technology. a Abbreviations: FabZ, β-hydroxyacyl-acyl carrier protein dehydra...
2 downloads 5 Views 1MB Size
J. Med. Chem. 2009, 52, 2465–2481

2465

Discovering Potent Inhibitors Against the β-Hydroxyacyl-Acyl Carrier Protein Dehydratase (FabZ) of Helicobacter pylori: Structure-Based Design, Synthesis, Bioassay, and Crystal Structure Determination† Lingyan He,‡,§ Liang Zhang,‡,§ Xiaofeng Liu,‡,§ Xianghua Li,§ Mingyue Zheng,§ Honglin Li,§ Kunqian Yu,§ Kaixian Chen,§ Xu Shen,*,§,| Hualiang Jiang,*,§,| and Hong Liu*,§ Center for Drug DiscoVery and Design, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China, School of Pharmacy, East China UniVersity of Science and Technology, Shanghai 200237, China ReceiVed December 10, 2008

The discovery of HpFabZ inhibitors is now of special interest in the treatment of various gastric diseases. In this work, three series of derivatives (compounds 3, 4, and 5) were designed, synthesized, and their biological activities were investigated as potential HpFabZ inhibitors in a two phased manner. First, we designed and synthesized two series of derivatives (3a-r and 4a-u) and evaluated the enzyme-based assay against HpFabZ. Five compounds (3i-k, 3m, and 3q) showed potential inhibitory activity, with IC50 values less than 2 µM. Second, a focused combinatorial library containing 280 molecules was designed employing the LD1.0 program. Twelve compounds (5a-l) were selected and synthesized. The activity of the most potent compound 5h (IC50 ) 0.86 µM) was 46 times higher than that of the hit 1. The high hit rate and the potency of the new HpFabZ inhibitors demonstrated the efficiency of the strategy for the focused library design and virtual screening. 1. Introduction Helicobacter pylori (H. pyloria) has recently been the subject of intense interest because it is a major causative factor for gastrointestinal illnesses such as chronic gastritis and peptic ulcers.1-5 Additionally, infection of H. pylori is also associated with adenocarcinomas and stomach lymphomas, increasing the risk of gastric cancer.6,7 Nevertheless, there is no effective therapy in eradicating H. pylori infection. Combination therapies employing one proton pump inhibitor (e.g., omeprazole) and two or three antibiotics (e.g., amoxicillin, clarithromycin, or tetracycline) have been used as preferred treatments.8-10 However, the multiple therapy regimens have not been very effective in the clinical setting because the bacterium is likely to develop resistance.11-14 Moreover, this treatment may disrupt the natural population of commensal microorganisms in the gastrointestinal tract, leading to undesired side effects such as diarrhea.15,16 Hence, a new level of treatment is needed to inactivate the key enzymes associated with the bacterial growth. Recently, the pathway of the type-II fatty acid biosynthesis system (FAS-II) has been particularly appreciated as an interest† PDB ID codes: 2GLL, 3DOY, 3DOZ, 3DP0, 3DP1, 3DP2, and 3DP3. * To whom correspondence should be addressed (X.S.) Phone: +8621-50806918. Fax: +86-21-50807088. E-mail: [email protected]. (H.J.) Phone: +86-21-50805873. Fax: +86-21-50807088. E-mail: hljiang@ mail.shcnc.ac.cn. (H.L.) Phone: +86-21-50807042. Fax: +86-21-50807088. E-mail: [email protected]. ‡ Authors equally contributed to this work. § Center for Drug Discovery and Design, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences. | School of Pharmacy, East China University of Science and Technology. a Abbreviations: FabZ, β-hydroxyacyl-acyl carrier protein dehydratase; Hp, Helicobacter pylori or H. pylori; FAS-I, type I fatty acid biosynthesis; FabD, malonyl-CoA:ACP transacylase; FabH, β-ketoacyl-acyl carrier protein synthase III; FabF, β-ketoacyl-acyl carrier protein synthase II; FabB, β-ketoacyl-acyl carrier protein synthase I; FabI, enoyl-acyl carrier protein reductase; FabA, β-hydroxyacyl-acyl carrier protein dehydratase; FabG, β-ketoacyl-ACP reductase I; ACP, the acyl carrier protein; CoA, acetyl coenzyme A; PfFabZ, Plasmodium falciparum FabZ.

Figure 1. The structures of compounds 1 and 2.

ing target for antibacterial agent discovery due to the major differences of the structural architectures and biological functions that exist between the plastid-associated enzymes of FASII found in most microorganisms and the enzymes involved in FAS-I for mammals and yeast.17-20 FAS-II consists of six key enzymes: FabD, FabH, FabF (or FabB), FabI, FabZ (or FabA), and FabG, all of which work in concert during the synthesis of fatty acid in vivo including the initiation and elongation phases. During the elongation cycle of FAS II, four consecutive reactions complete the extension of two carbons.21,22 In the third step of the elongation cycle, the dehydration of β-hydroxy-ACP to trans-2-acyl-ACP is catalyzed by FabZ or FabA. FabA is a bifunctional enzyme and is only found in Gram-negative bacteria with its partner, FabB, to participate in the formation of unsaturated fatty acids.23 However, FabZ has a ubiquitous distribution in the FAS II pathway and is a primary dehydratase that participates in the elongation cycles of both saturated and unsaturated fatty acid biosyntheses. Accordingly, FabZ presents itself as a suitable, yet unexplored target for the discovery of compounds against pathogenic microbes.22,23 So far, the only known small molecule inhibitors of FabZ are flavonoids discovered by random screening against Plasmodium falciparum FabZ (PfFabZ).24 Recently, we discovered two synthetic compounds (1 and 2, Figure 1) as inhibitors of H. pylori FabZ (HpFabZ) with IC50 values of 39.8 ( 0.35 µM

10.1021/jm8015602 CCC: $40.75  2009 American Chemical Society Published on Web 03/23/2009

2466 Journal of Medicinal Chemistry, 2009, Vol. 52, No. 8

and 47.6 ( 0.29 µM, respectively.25 In addition, we also determined the X-ray crystal structures of HpFabZ in complex with these inhibitors, which demonstrated the inhibitory mechanism of FabZ for the first time.25 In this study, on the basis of the structures and activities of compounds 1 and 2, and their interactive models with HpFabZ as well, we identified several small compounds that inhibit HpFabZ activity with IC50 values of 0.86-42.7 µM by employing a comprehensive approach integrating structure-based design, focused-library design, virtual screening, chemical synthesis, and bioassay. We approached anti-H. pylori therapeutic development employing structure-based design to identify small organic compounds as lead candidates. In the first phase of this study, we designed and synthesized two series of derivatives of compounds 1 and 2 according to the structural models of these inhibitors binding to HpFabZ. Five compounds showed high inhibition activities with IC50 values less than 2 µM, nearly 28 times more potent than the initial starting hits 1 and 2. Afterward, the crystal structures of the complexes of six compounds with the HpFabZ were determined. In the second phase, we adopted a strategy integrating focused combinatorial library design, virtual screening, chemical synthesis, and bioassay to identify more potent inhibitors based on the result of the first phase. Using the fragments taken from the HpFabZ inhibitors identified in the first phase, a focused combinatorial library containing 280 molecules was designed by employing the LD1.0 program26 on the basis of the crystal structures of the inhibitor-HpFabZ complexes. By using docking-based virtual screening targeting the binding site of HpFabZ, 12 compounds (5a-l) were selected for synthesis and bioassay. Eight compounds showed HpFabZ enzymatic inhibition activities with IC50 values of 0.86-42.7 µM. The inhibitory activity of the most potent compound increased 46 times more than that of the original compounds. All the inhibitors discovered in this study showed weak antibacterial activity, overcoming the shortcomings of the original inhibitors (compounds 1 and 2) due to improving the druglikeness of the designed compounds. The high hit rate and the high potency of the new HpFabZ inhibitors demonstrated the efficiency of the discovery strategy used in this study. In addition, the chemical entities reported in this study could be leading factors contributing to the discovery of new therapies against the FAS-II pathway.

2. Materials and Methods 2.1. Discovery Strategy. On the basis of our previously discovered HpFabZ inhibitors (compounds 1 and 2) and the crystal structures of the inhibitor-HpFabZ complexes, we started to design the new generation HpFabZ inhibitors to increase the enzymatic and antibacterial activities. The flowchart of the discovery strategy and procedure is outlined in Figure 2. In general, the inhibitor-designing process was conducted in a two-phase manner. During the phase I design, a series of derivatives of compounds 1 and 2 were designed and synthesized by a structure-based drug design approach according to the binding models of compounds 1 and 2 with HpFabZ. Several potent inhibitors were selected via an enzymatic activity assay, and the crystal structures of the complexes of the most potent inhibitors with HpFabZ were determined, which was the starting point for the phase II design. During the phase II design, a focused library containing 280 compounds was designed by using the fragments isolated from the inhibitors obtained in the phase I design as building blocks according to the inhibitor-HpFabZ binding models derived from the crystal structures. The structures in the focused library were docked

He et al.

Figure 2. Discovery strategy and experimental flowchart in this study.

back into the binding pockets of HpFabZ and were scored by virtual screening functions to select candidates for further synthesis and bioassay. Several new inhibitors were discovered in this phase of the design. Finally, the bacterial growth inhibitory activities of all the obtained HpFabZ inhibitors were assessed. The detailed experimental procedures of each step are described in the following sections. 2.1.1. Phase I Design. On the basis of the structural feature and their interaction models with HpFabZ of compounds 1 and 2, 39 new analogues (3a-r and 4a-u, Tables 1 and 2) were designed and synthesized in the first phase. Keeping the pyridine ring of compound 1, we obtained compounds 3a-d by the removal of the hydroxyl group or bromide or by introducing methoxyl groups to the phenyl ring. Keeping the 2,4-dihydroxy3,5-dibromo phenyl ring and replacing the pyridine ring with the phenyl ring, substituted phenyl ring, methyl, naphthalene ring, or various heterocycles, we obtained compounds 3e-r. On the basis of the structural features of compound 2, we designed compounds 4a-u (Table 2). By changing the substituent groups of the substitutional phenyl ring R3 of compound 2, we got compounds 4a-c. Compounds 4d-r were obtained by introducing various electronic, hydrophobic groups to the ortho-, meta-, or para- position of the unsubstituted phenyl ring R4 of compound 2 or displacing the R4 group with the naphthalin ring. Compounds 4s-u were obtained by displacing the methoxy group with three different amino groups. 2.1.2. Phase II Design. Before phase II design, the X-ray crystal structures of HpFabZ in complex with the inhibitors produced from phase I design were determined, which provide inhibitor-enzyme interaction models for further inhibitor design. A focused library was designed using the structural fragments isolated by the above discovered inhibitors according to the inhibitor-enzyme interaction models. The LD1.0 program26,27 was used during the focused library design. The inhibitor-enzyme interaction models indicate that the HpFabZ inhibitors can be divided into three parts (Figure 3A): part A is located in the large pocket (site A), part B interacts with the small pocket of HpFabZ (site B), and part L is a linker between A and B, interacting with the “saddle” pocket between sites A and B, with a length of 5-6 Å (Figure 3A, middle image). From fragments of HpFabZ inhibitors discovered above and previously,25,28 the LD1.0 program optimized 14 fragments for part A, 5 fragments for part B, and 4 linkers for part L. The chemical structures of the fragments are shown in Figure 4. Then LD1.0 connected these fragments together according to the structure

Inhibitors Against the FabZ of Helicobacter pylori

Journal of Medicinal Chemistry, 2009, Vol. 52, No. 8 2467

Table 1. Chemical Structures, HpFabZ Inhibitory Activities, and Antibacterial Activity of Compounds 1 and 3a-r

a Values are means of three determinations and deviation from the mean is 100 µg/mL. Among the compounds, 3m, 3q,

2472 Journal of Medicinal Chemistry, 2009, Vol. 52, No. 8

He et al.

Figure 5. Schematic picture of the superposition of HpFabZ-compounds complex. Two molecules of each compound (except compound 3m) bind to one hexamer of HpFabZ with two distinct interaction models. HpFabZ dimer A/B, C/D, compound 1, compounds 3i, 3j, 3k, 3m, 3n, and 3q are colored yellow/magenta, blue/orange, cyan, and green, respectively. The active-site cavities are produced with the program MOLE52 and shown in dark-gray surface representations. Table 3. Chemical Structures, HpFabZ Inhibitory Activities and Antibacterial Activity of The 12 Compounds Selected From The Focused Library Using Virtual Screening

a

Values are the means of three determinations and deviation from the mean is